Language selection

Search

Patent 2982963 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2982963
(54) English Title: CHIMERIC ANTIGEN RECEPTOR SPECIFIC FOR TUMOR CELLS
(54) French Title: RECEPTEUR D'ANTIGENE CHIMERIQUE SPECIFIQUE AUX CELLULES DE TUMEUR
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 19/00 (2006.01)
  • A61P 35/00 (2006.01)
  • C07K 14/705 (2006.01)
  • C07K 16/28 (2006.01)
  • C07K 16/30 (2006.01)
  • C07K 16/46 (2006.01)
  • C12N 5/10 (2006.01)
  • G01N 33/574 (2006.01)
(72) Inventors :
  • BOSIO, ANDREAS (Germany)
  • ECKARDT, DOMINIK (Germany)
  • HARDT, OLAF (Germany)
  • DZIONEK, ANDRZEJ (Germany)
  • TOMIUK, STEFAN (Germany)
  • KOLLET, JUTTA (Germany)
(73) Owners :
  • MILTENYI BIOTEC B.V. & CO. KG (Germany)
(71) Applicants :
  • MILTENYI BIOTEC GMBH (Germany)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued: 2024-02-06
(22) Filed Date: 2017-10-19
(41) Open to Public Inspection: 2018-04-20
Examination requested: 2022-06-23
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): No

(30) Application Priority Data:
Application No. Country/Territory Date
16194708.0 European Patent Office (EPO) 2016-10-20

Abstracts

English Abstract

The present invention is directed to ligand like a chimeric antigen receptor (CAR), comprising an antigen binding domain specific for one or more antigens selected from the group consisting of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8; cell populations expressing such CARs and the use of the cell populations for cancer therapy.


French Abstract

La présente invention concerne un ligand, comme un récepteur antigénique chimérique (CAR), qui comprend un domaine dattachement pour antigène spécifique à au moins un antigène sélectionné dans le groupe composé de CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 et TSPAN8. Des populations de lymphocytes exprimant de tels CAR et lutilisation de ces populations dans la cancérothérapie sont aussi décrites.

Claims

Note: Claims are shown in the official language in which they were submitted.


38
CLAIMS:
1) A chimeric antigen receptor (CAR), comprising an antigen binding domain
specific for
CD318 in combination with an antigen binding domain specific for CLA.
2) The chimeric antigen receptor (CAR) according to claim 1, wherein the
CAR comprises
a transmembrane domain and/or an intracellular signaling domain and wherein
the antigen
binding domains are conjugated to the same or a different transmembrane domain
and/or
intracellular signaling domain.
3) The chimeric antigen receptor (CAR) according to claim 2, wherein the
transmembrane
domain comprises 4-1BB, CD8 and/or CD28; and the intracellular signaling
domain comprises
one or more of CD28, CD137 and CD3zeta.

Description

Note: Descriptions are shown in the official language in which they were submitted.


1
CHIMERIC ANTIGEN RECEPTOR SPECIFIC FOR TUMOR CELLS
Field of invention
The present invention relates to the use of ligands comprising antigen binding
domains specific
for certain antigens, like chimeric antigen receptors (CAR) and/or engineered
cells provided with
such ligands for treatment of human cancer.
Background
Cancer is a broad group of diseases involving unregulated cell growth. In
cancer, cells divide and
grow uncontrollably, forming malignant tumors, and invading nearby parts of
the body. The cancer
may also spread to more distant parts of the body through the lymphatic system
or bloodstream.
There are over 200 different known cancers that affect humans. Whereas good
treatment options
are available for many cancer types, others still represent unmet medical
needs.
The technology of chimeric antigen receptor (CAR) may provide a promising
approach for
adoptive cell immunotherapy for cancer. Commonly, CARs comprise a single chain
fragment
variable (scFv) of an antibody specific for a tumor associated antigen (TAA)
coupled via hinge
and transmembrane regions to cytoplasmic domains of T-cell signaling
molecules. For example,
well known lymphocyte activation moieties include a T-cell costimulatory (e.g.
CD28, CD137,
0X40, ICOS, and CD27) domain in tandem with a T-cell triggering (e.g. CD3c)
moiety. The CAR-
mediated adoptive immunotherapy allows CAR-grafted cells to directly recognize
the TAAs on
target tumor cells in a non-HLA-restricted manner.
Paramount for immunotherapy for cancer based on CAR is the selection of
antigens specific for
the respective tumor cells. Object of the invention was to provide such
antigens specific for cancer
cells, especially for pancreas cancer cells in order to engineer killer cells
which then kill/lyse
cancer cells without attacking non-tumor cells.
Date Recue/Date Received 2022-11-04

2
Summary of the invention
It has been found that a distinct group of cell surface antigens is expressed
on several human cancer
cells, especially on human pancreas cancer cells, but not or to a lower level
on non-malignant cells.
Accordingly, these antigens (also referred to as "markers") can be used to
identify and/or mark
and/or destroy and/or disable escape mechanisms of such cancer cells via
ligands that specifically
bind to the markers.
Therefore, the invention relates to a ligand, comprising an antigen binding
domain specific for one
or more antigens characterized in that the ligand is a chimeric antigen
receptor (CAR), comprising
an antigen binding domain specific for one or more antigens selected from the
group consisting of
CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8.
Another object of the invention are methods of binding a cancer cell with a
ligand comprising an
antigen binding domain specific for one or more antigens selected from the
group consisting of
CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8.
The ligands as further disclosed may be an antibody or a CAR or an engineered
cell expressing at
least one such ligands.
Another objects of the invention are populations of engineered cells
expressing at least one of said
ligands, pharmaceutical compositions comprising the population of engineered
cells and/or the use
of the population of engineered cells or the pharmaceutical composition for
treatment of human
cancer.
Brief description of the drawings
Fig. 1 shows the general structure of a CAR capable of recognizing a specific
target
Fig. 2 shows variant of the general structures of the CARs according to the
invention. Fig. 2A
shows a single CAR; Fig. 2B shows a split CAR; Fig. 2C shows a tandem CAR;
Fig. 2D
shows multiple CARS from one vector
Fig. 3 shows the expression of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318
and TSPAN8
on xenotransplanted human pancreatic cancer cells
Date Recue/Date Received 2022-11-04

3
Fig. 4 shows the expression of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318
and TSPAN8
on primary human pancreatic cancer cells and healthy tumor infiltrating cells
Fig 5 shows the expression of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and
TSPAN8
on primary human pancreatic cancer sections
Fig. 6 A shows the percentage of cells expressing LNGFR on the surface as a
readout of
transfection efficacy and construct expression. Fig. 6 B shows the percentage
of cells
expressing the CAR construct on the surface
Fig. 7 shown the killing efficacy of CAR T cells towards pancreatic cancer
cells.
Fig. 8 shows in (A) the gating and in (B) the co-expression of the markers
Detailed description of the invention
In a first embodiment of the invention, the ligand comprises at least two
different antigen binding
domains specific for at least two different antigens selected from the group
consisting of CLA,
CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8. For example, the ligand
may
comprise antigen binding domains specific for CLA and CD66c or specific for
CLA and TSPAN8.
In a preferred method of binding a cancer cell, the cancer cell (or population
of cancer cells) is
bound with a ligand comprised at least two different antigen binding domains
specific for at least
two different antigens selected from the group consisting of CLA, CD142, CD73,
CD49c, CD66c,
CD104, CD318 and TSPAN8. Again, the ligand may comprise antigen binding
domains specific
for CLA and CD66c or specific for CLA and TSPAN8.
Definitions
The term "tumor" is known medically as a neoplasm. Not all tumors are
cancerous; benign tumors
do not invade neighboring tissues and do not spread throughout the body.
The term "cancer" is known medically as a malignant neoplasm. Cancer is a
broad group of
diseases involving unregulated cell growth. In cancer, cells (cancerous cells)
divide and grow
uncontrollably, forming malignant tumors, and invading nearby parts of the
body. The cancer may
also spread to more distant parts of the body through the lymphatic system or
bloodstream.
Date Recue/Date Received 2022-11-04

4
The term "isolated" means altered or removed from the natural state. For
example an isolated
population of cells means an enrichment of such cells and separation from
other cells which are
normally associated in their naturally occurring state with said isolated
cells. An isolated
population of cells means a population of substantially purified cells which
is a homogenous
population of cells.
The terms "specifically binds" or "specific for" with respect to an antigen-
binding domain of a
ligand like an antibody, of a fragment thereof or of a CAR refer to an antigen-
binding domain
which recognizes and binds to a specific antigen, but does not substantially
recognize or bind other
molecules in a sample. An antigen-binding domain that binds specifically to an
antigen from one
species may bind also to that antigen from another species. This cross-species
reactivity is not
contrary to the definition of that antigen-binding domain as specific. An
antigen-binding domain
that specifically binds to an antigen may bind also to different allelic forms
of the antigen (allelic
variants, splice variants, isoforms etc.). This cross reactivity is not
contrary to the definition of that
antigen-binding domain as specific.
The terms "engineered cell" and "genetically modified cell" as used herein can
be used
interchangeably. The terms mean containing and/or expressing a foreign gene or
nucleic acid
sequence which in turn modifies the genotype or phenotype of the cell or its
progeny. Especially,
the terms refers to cells, preferentially T cells which are manipulated by
recombinant methods well
known in the art to express stably or transiently peptides or proteins which
are not expressed in
these cells in the natural state. For example, T cells are engineered to
express an artificial construct
such as a chimeric antigen receptor on their cell surface. For example, the
sequences encoding the
CAR may be delivered into cells using a retroviral or lentiviral vector.
The amino acid sequences given in SEQ ID NO:! - 32, respectively (in the one-
letter code of
amino acids) shall refer to all constellations of the respective amino acid
sequence which retains
the intended function of the respective amino acid sequence as defined.
Therefore, all variants of
the amino acid sequences defined in the sequence listings having a sequence
identity of at least
70%, or at least 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% at the amino acid
sequence level
are included in the scope of the present invention. In the context of the
present invention, "sequence
Date Recue/Date Received 2022-11-04

5
identity" may be determined using pairwise alignments using alignments
programs for amino acid
sequences well known to the art.
T cells or T lymphocytes are a type of lymphocyte that play a central role in
cell-mediated
immunity. They can be distinguished from other lymphocytes, such as B cells
and natural killer
cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell
surface. There are several
subsets of T cells, each with a distinct function.
T helper cells (TH cells) assist other white blood cells in immunologic
processes, including
maturation of B cells into plasma cells and memory B cells, and activation of
cytotoxic T cells and
macrophages. These cells are also known as CD4+ T cells because they express
the CD4
glycoprotein on their surface. Helper T cells become activated when they are
presented with
peptide antigens by MHC class H molecules, which are expressed on the surface
of antigen-
presenting cells (APCs). Once activated, they divide rapidly and secrete small
proteins called
cytokines that regulate or assist in the active immune response. These cells
can differentiate into
one of several subtypes, including TH1, TH2, TH3, TH17, Th9, or TFH, which
secrete different
cytokines to trigger a different type of immune response. Signaling from the
APC directs T cells
into particular subtypes.
Cytotoxic T cells (Tc cells, or CTLs) destroy infected cells and tumor cells,
and are also implicated
in transplant rejection. These cells are also known as CD8+ T cells since they
express the CD8
glycoprotein at their surface. These cells recognize their targets by binding
to antigen associated
with MHC class I molecules, which are present on the surface of all nucleated
cells.
Memory T cells are a subset of antigen-specific T cells that persist long-term
after an infection has
resolved. They quickly expand to large numbers of effector T cells upon re-
exposure to their
cognate antigen, thus providing the immune system with "memory" against past
infections.
Memory T cells comprise three subtypes: central memory T cells (Tcm cells) and
two types of
effector memory T cells (Tim cells and TEMRA cells). Memory cells may be
either CD4+ or CD8+.
Memory T cells typically express the cell surface molecule CD45RO.
Date Recue/Date Received 2022-11-04

6
Regulatory T cells (Treg cells), formerly known as suppressor T cells, are
crucial for the
maintenance of immunological tolerance. Their major role is to shut down T
cell-mediated
immunity toward the end of an immune reaction and to suppress auto-reactive T
cells that
escaped the process of negative selection in the thymus. Two major classes of
CD4+ Leg cells
have been described ¨ Foxp3+ Tin cells and Foxp3- Treg cells.
Natural killer T cells (NKT cells) bridge the adaptive immune system with the
innate immune
system. Unlike conventional T cells that recognize peptide antigens presented
by major
histocompatibility complex (MHC) molecules, NKT cells recognize glycolipid
antigen presented
by a molecule called CD1d. Once activated, these cells can perform functions
ascribed to both Th
and 11 cells (i.e., cytokine production and release of cytolytic/cell killing
molecules).
Immunotherapy is a medical term defined as the "treatment of disease by
inducing, enhancing, or
suppressing an immune response". Immunotherapies designed to elicit or amplify
an immune
response are classified as activation immunotherapies, while immunotherapies
that reduce or
suppress are classified as suppression immunotherapies. Cancer immunotherapy
as an activating
immunotherapy attempts to stimulate the immune system to reject and destroy
tumors. Adoptive
cell transfer uses cell-based, such as T cell-based cytotoxic responses to
attack cancer cells. T cells
that have a natural or genetically engineered reactivity to a patient's cancer
are generated in vitro
and then transferred back into the cancer patient.
The term "biomarker" or "marker" is widespread in the art and may broadly
denote a biological
molecule and/or a detectable portion thereof (e.g. a nucleic acid, a peptide
or a lipid such as a
glycolipid) whose qualitative and/or quantitative evaluation in an individual
is predictive or
informative (e.g., predictive, diagnostic and/or prognostic) with respect to
one or more aspects of
the individual's phenotype and/or genotype. E.g. the biomarker is predictive
or informative with
respect to the outcome for chemotherapeutic treatment of a cancer in an
individual. A biomarker
is expressed ("expression of the biomarker") if the biomarker is detectable
with methods known
in the art. Therefore expression of biomarkers encompasses not only expression
at nucleic acid
level (DNA and/or RNA) and protein level but also expression (presence) of
other biological
structures on or in the cells such as glycolipids or the activity of a
protein.
Date Recue/Date Received 2022-11-04

7
The term "target" as used herein refers to an antigen or epitope associated
with a cell that should
be recognized specifically by an antigen binding domain, e.g. an antigen
binding domain of an
antibody or of a CAR. The antigen or epitope for antibody recognition can be
bound to the cell
surface but also be secreted, part of the extracellular membrane, or shed from
the cell.
The term "antibody" as used herein refers to polyclonal or monoclonal
antibodies and fragments
thereof, which can be generated by methods well known to the person skilled in
the art. The
antibody may be of any species, e.g. mice, rat, sheep, human. For therapeutic
purposes, if non-
human antigen binding fragments are to be used, these can be humanized by any
method known
in the art. The antibodies may also be modified antibodies (e.g. oligomers,
reduced, oxidized and
labeled antibodies).
The term "killer cell" as used herein refers to a cell that can kill/lyse
another cell, e.g. a cancer cell.
Most frequently, T cells, NK cells, dendritic cells and macrophages can be
used as killer cells.
The term "engineered killer cell" as used herein refers to a killer cell that
is genetically modified
to allow for the specific killing of a target cell, e.g. a cell modified with
a CAR against a target to
kill tumor cell expressing the respective target.
Chimeric Antigen Receptor (CAR)
The chimeric antigen receptor (CAR) according to the invention may comprise an
antigen binding
domain conjugated to a transmembrane domain and/or a intracellular signaling
domain, as shown
by way of example in Fig. 1.
In a first embodiment of the invention, the ligand is a chimeric antigen
receptor (CAR), comprising
an antigen binding domain specific for one or more antigens selected from the
group consisting of
CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8.
In a second embodiment of the invention, the ligand is a CAR, comprising an
antigen binding
domain specific for CLA in combination with one or more antigens selected from
the group
consisting of CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8.
Date Recue/Date Received 2022-11-04

8
In a third embodiment of the invention, the ligand is a CAR, comprising an
antigen binding domain,
an transmembrane domain and/or an intracellular signaling domain and
comprising at least two
antigen binding domains specific for two different antigens selected from the
group consisting of
CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8 are conjugated to the
same or
a different transmembrane domain and/or intracellular signaling domain.
In another embodiment of the invention, the antigen binding domain of a CAR
binds a hapten that
is coupled to a polypeptide ("haptenylated" polypeptide), wherein the
polypeptide may bind to a
tumor associated antigen. Such CARs are for example disclosed in US9233125B2
and are known
in the art as "anti-tag" CAR. Similar, the extracellular part of the CAR of
the invention may
comprise a linker/label epitope (LLE) binding domain as antigen binding domain
that binds to a
linker/label epitope (LLE) that is part of a target cell binding molecule Such
"anti-LLE CARs" are
disclosed in the European patent application no. EP16196487.9. Both types of
CARs are universal
and/or adaptable CAR. Both the hapten(s) and the LLE are "tags" that are
coupled directly or
indirectly to a polypeptide (the tagged polypeptide), wherein the polypeptide
may bind to a tumor
associated antigen expressed on the (cell) surface of a target cell.
In this embodiment, the ligand comprises an antigen binding domain specific
for one or more
antigens characterized in that the ligand is a chimeric antigen receptor
(CAR), comprising an anti
tag binding region which can bind to a tag which is coupled to an antigen
binding domain specific
for one or more antigens selected from the group consisting of CLA, CD142,
CD73, CD49c,
CD66c, CD104, CD318 and TSPAN8. Suitable tags are for example, but not limited
to, Biotin,
other haptens, FITC or other fluorochrome molecules, FLAG, HIS, YOL MYC,
Dextran, FcR,
antibody-isotypes, artificially engineered epitopes, FAB or FAB2 binders.
The transmembrane domain of the CAR may comprise a sequence of the
transmembrane domains
of 4-1BB, CD8 and/or CD28; and the intracellular signaling domain comprises a
sequence of the
intracellular signaling domains of one or more of CD28, CD137 and CD3zeta.
In a preferred variant of this embodiment, the chimeric antigen receptor (CAR)
comprises an
antigen binding domain specific for CLA without an additional antigen binding
domain or
Date Recue/Date Received 2022-11-04

9
additional CAR, wherein the antigen binding domain is conjugated to one
transmembrane domains
and one or more signaling domains. This variant is shown by way of example in
Fig. 2 A.
In a second variant of the invention, the chimeric antigen receptor (CAR)
comprises at least two
antigen binding domains specific for two or more antigens selected from the
group consisting of
CLA (cutaneous lymphocyte antigen), CD142, CD73, CD49c, CD66c, CD104, CD318
and
TSPAN8, wherein the antigen binding domains are conjugated to different
transmembrane
domains and/or signaling domains. This variant is shown by way of example in
Fig. 2 b.
In a third variant of the invention, the chimeric antigen receptor (CAR)
comprises at least two
antigen binding domains specific for two or more antigens selected from the
group consisting of
CLA (cutaneous lymphocyte antigen), CD142, CD73, CD49c, CD66c, CD104, CD318
and
TSPAN8, wherein the antigen binding domains are conjugated to the same (one)
transmembrane
domain and signaling domains. This variant is shown by way of example in Fig.
2 c.
In a forth variant of the invention, the chimeric antigen receptor (CAR)
comprises at least two
antigen binding domains specific for two or more antigens selected from the
group consisting of
CLA (cutaneous lymphocyte antigen), CD142, CD73, CD49c, CD66c, CD104, CD318
and
TSPAN8, wherein the antigen binding domains are conjugated to different
transmembrane
domains and signaling domains and the antigen binding domains origin from one
vector. This
variant is shown by way of example in Fig. 2 d.
CLA is the cutaneous lymphocyte-associated antigen (CLA), a specialized
glycoform of P-selectin
glycoprotein ligand-1 (PSGL-1). It serves as a ligand for selectins, including
CD62E (ELAM-1)
and CD62L (LECAM-1). CLA is a unique skin-homing receptor and is predominantly
found on a
minor subset of human T cells that infiltrate the skin. This post-
translational modification of
PSGL-1 is thought to serve as a mechanism to regulate tissue-specific homing
of CD4+ and CD8+
memory/effector T cells from peripheral blood to the skin, which plays an
essential role during
many inflammatory and certain malignant skin diseases.
Date Recue/Date Received 2022-11-04

10
In peripheral blood, CLA is not only found on skin-homing memory/effector T
cells, but is also
found to be expressed on memory/effector B cells, NK cells, blood dendritic
cells, and on
monocytes. CLA is furthermore found on Langerhans cells in the skin.
In order to enhance the specific recognition of cancer cells, the chimeric
antigen receptor (CAR)
may comprise an antigen binding domain specific for CLA in combination with
one or more (like
two, three or four) antigens selected from the group consisting of CD142,
CD73, CD49c, CD66c,
CD 104, CD318 and TSPAN8. Preferred combinations specific for pancreas cancer
are CLA with
TSPAN8 and CLA with CD66c.
The antigen binding domain of said CAR may comprise, for example, full length
heavy chain, Fab
fragments, single chain Fv (scFv) fragments, divalent single chain antibodies
or diabodies, each of
which are specific for one or more of the target antigens CLA, CD142, CD73,
CD49c, CD66c,
CD104, CD318 and TSPAN8.
The antigen binding domain of said CAR may comprise the amino acid sequences
of SEQ ID
NO:1 and SEQ ID NO:2. The relevant sites causing specificity for antigen
binding are the CDRs
according to the IMGT (the international ImMunoGeneTics information system for

immunoglobulins or antibodies) definition which are underlined in the
sequence. The antigen
binding domain of said CAR may comprise a scPv comprising the amino acid
sequence of SEQ
ID NO:17 or SEQ ID NO:18.
The present invention also encompasses nucleic acids (DNA or RNA) constructs
comprising
sequences encoding for amino acids sequences of a CAR specific for the
disclosed markers.
In one embodiment of the invention a DNA construct (vector, plasmid) is
generated encoding for
a CAR specific for the disclosed markers. A nucleic acid sequence encoding for
an antigen binding
domain specific for the disclosed markers is fused at least to a nucleic acid
sequence encoding a
transmembrane domain and subsequent a nucleic acid sequence encoding a
intracellular domain.
The construction of such expression vectors can be performed by recombinant
methods well
known in the art. Alternatively, the nucleic acid sequences can be produced
synthetically.
Date Recue/Date Received 2022-11-04

11
Alternatively, the CAR may be composed of further parts such as a linker
and/or hinge and/or may
be composed as di- or multi-chain CAR as described below.
As shown in general in Fig. 1 and 2, a CAR may comprise an extracellular
domain comprising the
antigen binding domain, a transmembrane domain and an intracellular signaling
domain. The
extracellular domain may be linked to the transmembrane domain by a linker.
The extracellular
domain may also be linked to a signal peptide.
A "signal peptide" refers to a peptide sequence that directs the transport and
localization of the
protein within a cell, e.g. to a certain cell organelle (such as the
endoplasmic reticulum) and/or the
cell surface.
An "antigen binding domain" refers to the region of the CAR that specifically
binds to an antigen
(and thereby is able to target a cell containing an antigen). The CARs of the
invention may
comprise one or more antigen binding domains. Generally, the antigen binding
domain on the
CAR are extracellular. The antigen binding domain may comprise an antibody or
a fragment
thereof. The antigen binding domain may comprise, for example, full length
heavy chain, Fab
fragments, single chain Fv (scFv) fragments, divalent single chain antibodies
or diabodies. Any
molecule that binds specifically to a given antigen such as affibodies or
ligand binding domains
from naturally occurring receptors can be used as an antigen binding domain.
Often the antigen
binding domain is a scFv. Normally, in a scFv the variable portions of an
immunoglobulin heavy
chain and light chain are fused by a flexible linker to form a scFv. Such a
linker may be for example
the "(G4/S1)3-linker".
In some instances, it is beneficial for the antigen binding domain to be
derived from the same
species in which the CAR will be used in. For example, if it is planned to use
it therapeutically in
humans, it may be beneficial for the antigen binding domain of the CAR to
comprise a human or
humanized antibody or fragment thereof. Human or humanized antibodies or
fragments thereof
can be made by a variety of methods well known in the art.
Date Recue/Date Received 2022-11-04

12
"Spacer" or "hinge" as used herein refers to the hydrophilic region which is
between the antigen
binding domain and the transmembrane domain. The CARs of the invention may
comprise an
extracellular spacer domain but is it also possible to pass such a spacer. The
spacer may include
Fc fragments of antibodies or fragments thereof, hinge regions of antibodies
or fragments thereof,
CH2 or CH3 regions of antibodies, accessory proteins, artificial spacer
sequences or combinations
thereof. A prominent example of a spacer is the CD8alpha hinge.
The transmembrane domain of the CAR can be derived from any desired natural or
synthetic
source for such domain. If the source is natural the domain may be derived
from any membrane-
bound or transmembrane protein. The transmembrane domain may be derived for
example from
CD8alpha or CD28.
The cytoplasmic domain or the intracellular signaling domain of the CAR of the
invention is
responsible for activation of at least one of the normal effector functions of
the immune cell in
which the CAR is expressed. "Effector function" means a specialized function
of a cell, e.g. in a T
cell an effector function may be cytolytic activity or helper activity
including the secretion of
cytokines. The intracellular signaling domain refers to the part of a protein
which transduces the
effector function signal and directs the cell expressing the CAR of the
invention to perform a
specialized function. The intracellular signaling domain may include any
complete or truncated
part of the intracellular signaling domain of a given protein sufficient to
transduce the effector
function signal.
Prominent examples of intracellular signaling domains for use in the CARs
include the
cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act
in concert to initiate
signal transduction following antigen receptor engagement.
Generally, T cell activation can be mediated by two distinct classes of
cytoplasmic signaling
sequence, firstly those that initiate antigen-dependent primary activation
through the TCR
(primary cytoplasmic signaling sequences) and secondly those that act in an
antigen-independent
manner to provide a secondary or co- stimulatory signal (secondary cytoplasmic
signaling
sequences).
Date Recue/Date Received 2022-11-04

13
Primary cytoplasmic signaling sequences that act in a stimulatory manner may
contain ITAMs
(immunoreceptor tyrosine-based activation motifs signaling motifs).
Examples of ITAM containing primary cytoplasmic signaling sequences often used
in CARs
derived from TCR zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon,
CD5,
CD22, CD79a, CD79b, and CD66d.
The cytoplasmic domain of the CAR can be designed to comprise the CD3-zeta
signaling domain
by itself or combined with any other desired cytoplasmic domain(s). The
cytoplasmic domain of
the CAR can comprise a CD3 zeta chain portion and a costimulatory signaling
region. The
costimulatory signaling region refers to a part of the CAR comprising the
intracellular domain of
a costimulatory molecule. A costimulatory molecule is a cell surface molecule
other than an
antigen receptor or their ligands that is required for an efficient response
of lymphocytes to an
antigen. Examples for costimulatory molecule are CD27, CD28, 4-1BB (CD137),
0X40, CD30,
CD40, PD-1, ICOS, lymphocyte function-associated antigen- 1 (LFA-1), CD2, CD7,
LIGHT,
NKG2C, B7-H3.
The cytoplasmic signaling sequences within the cytoplasmic signaling part of
the CAR may be
linked to each other in a random or specified order. A short oligo- or
polypeptide linker, which is
preferably between 2 and 10 amino acids in length, may form the linkage. A
prominent linker is
the glycine- serine doublet.
As an example, the cytoplasmic domain may comprise the signaling domain of CD3-
zeta and the
signaling domain of CD28. In another example the cytoplasmic domain may
comprise the
signaling domain of CD3-zeta and the signaling domain of CD27. In an further
example, the
cytoplasmic domain may comprise the signaling domain of CD3-zeta, the
signaling domain of
CD28, and the signaling domain of CD27.
The CAR of the invention may be designed to comprise any portion or part of
the above-mentioned
domains as described herein, especially in the variants shown in Fig. 2 a-d.
The specificity of the
CAR of the invention mediated by the antigen binding domain is for one or more
of the antigens
Date Recue/Date Received 2022-11-04

14
CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8 all other domains
necessary
to construct a functional CAR may be chosen from the options mentioned above
or which are well
known to the person skilled in the art.
engineered cells expressing the ligand
In another embodiment of the invention, the ligand is an engineered cell (or a
population thereof),
expressing at least one antigen binding domain specific for one or more
antigens selected from the
group consisting of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8.
In a preferred embodiment, the population of engineered cells express an
ligand like an chimeric
antigen receptor (CAR) comprising an antigen binding domain specific for CLA
in combination
with one or more CARs and/or antigens selected from the group consisting of
CD142, CD73,
CD49c, CD66c, CD104, CD318 and TSPAN8.
The population of engineered cells may consist of T cells, macrophages or NK
cells. The
population of engineered cells may be are expanded to an therapeutically
effective amount of cells
before use in said immunotherapy.
To generate cells expressing the one or more CAR of the invention (including
the variants), a DNA
construct encoding the CAR of the invention can be transfected or transduced
into a host cell by
methods well known in the art (e.g. viral-based systems, physical methods,
biological methods,
chemical methods). Regardless of the methods used to integrate the nucleic
acid encoding the CAR
of the invention in the host cell, as a result the host cell expresses a CAR
which is specific for the
markers as disclosed.
In one embodiment of the invention, the engineered cells are isolated
(enriched or separated) after
the transfection/transduction process for generating such an engineered cell
from non-
transfected/transduced cells by methods well known in the art, e.g.
fluorescent based separating
technologies such as PACS or magnetic cell separation methods such as MACS .
Date Recue/Date Received 2022-11-04

15
In another embodiment of the invention a source of immune cells,
preferentially T cells is obtained
from a subject. Immune cells, preferentially T cells can be obtained from a
variety of sources such
as peripheral blood mononuclear cells (PMBCs), bone marrow, lymph node tissue,
cord blood or
thymus tissue. For enrichment of these cells methods well known in the art can
be used such as
centrifugation through a Ficoll' or PERCOLL
gradient or positive/negative selection
techniques such as fluorescent sorting (e.g. FACSsort) or magnetic sorting
(e.g. MACRO).
For example, T cells of a blood sample of a subject are magnetically labelled,
for example with a
magnetic bead coupled to antibodies specific for CD4 and for CD8 or
alternatively CD62L,
respectively, washed, magnetically enriched and collected. Then these T cells
may be engineered
to express the antigens as disclosed or the preferred combination of antigens
on their cell surface.
In one embodiment of the invention the isolated/enriched engineered T cells
expressing an antigens
as disclosed or the preferred combination of antigens prior or after genetic
modification can be
activated and expanded to increase amount of engineered T cells generally
using methods well
known in the art, for example polyclonal stimulation with anti-CD3/anti-CD28
beads or anti-
CD3/anti-CD28 nanomatrices (as disclosed in EP2711418A1). Preferentially, said
amount of
engineered T cells is increased to a therapeutic effective amount.
In one embodiment of the invention a cell expressing the CAR of the invention
is generated. The
RNA encoding the CAR of the invention can be transfected or transduced into a
host cell by
methods well known in the art (e.g. viral-based systems, physical methods,
biological methods,
chemical methods). In general, such an "RNA-engineered cell" is disclosed in
detail in
W02013/040557. Regardless of the methods used to integrate the RNA encoding
the CAR of the
invention in the host cell, as a result the host cell expresses a CAR which is
specific for an antigen
as disclosed or the preferred combination of antigens. Using "RNA-engineered
cells" lead to the
fact that the CAR is expressed for a limited time in the cell (transient
expression).
In one embodiment of the invention, the engineered cells are generated
automatically in a closed
cell culture system. Such process may comprises the steps:
a) providing a cell sample
Date Recue/Date Received 2022-11-04

16
b) preparation of the cell sample by centrifugation
c) magnetic separation of the cell, preferentially T cells, T cell subsets or
T cell progenitors
d) activation of the enriched cells, preferentially T cells, T cell subsets or
T cell progenitors using
modulatory agents
e) genetically modifying the cells, preferentially T cells, T cell subsets or
T cell progenitors to
express one or more CARs as disclosed or the preferred combination of
CARs/antigens
f) expansion of the genetically modified T cells, T cell subsets or T cell
progenitors
in a cultivation chamber
g) washing of the cultured cells, preferentially T cells, T cell subsets or T
cell progenitors.
All these steps may be performed in a closed and sterile system.
The process is especially suited for preparing gene modified cells,
preferentially T cells, T cell
subsets or T cell progenitors wherein the enriched cells, preferentially T
cells, T cell subsets or T
cell progenitors are gene modified by using viral and/or non-viral vectors.
Any of these steps may
be multiplied, omitted or may occur in a different order. In a variant of the
invention, the
modulatory agents are selected from agonistic antibodies and / or cytolcines.
As closed and sterile system for cell modification, the fully automated cell
processing device
CliniMACS Prodigy and associated tubing sets (Miltenyi Biotec GmbH, Germany)
may be used
(W02009/072003). This closed system meets the requirements of GMP-grade
processing of
almost any kind of cellular products and may allow reducing clean room
requirements, improve
technology transfer and harmonization of cell manufacturing processes. It has
been developed to
fully automate and standardize the manufacturing process of cellular
therapeutic agents. The
instrument can perform sample loading, cell washing, density-based cell
separations including
erythrocyte reduction and plasma harvesting, magnetic separation, cell
activation, cell
modification (transduction), cell culture, and final product formulation.
Thus enabling the flexible integration of process modules ("steps") in a
closed, automated and safe
GMP compliant workflow reproducing a complex desired biological process.
Date Recue/Date Received 2022-11-04

17
In one embodiment of the invention, the genetically modified cells express one
of the targets. To
circumvent killing among the genetically modified cell population, this target
is temporarily or
permanently knocked down or knocked out on the killer cells. Temporal or
permanent knock down
or knock out of expression can be induced by methods well known in the art,
such as siRNA for
temporal knock down or the CRISPR system for permanent knock out To inhibit
target expression
using these methods, this can be achieved by directly targeting the whole gene
encoding for the
target, parts of the gene, e.g. specific exons, the promotor region, or
controlling genes, such as
transcription factors. In the case of target structures representing
glycostructures, such as CLA,
this can also be achieved by altering the glycosylation site on the backbone
protein or one or more
of the enzymes catalyzing the glycosylation.
Methods of use
Another embodiment of the invention is a method of binding a cancer cell with
a ligand comprising
at least two different antigen binding domains specific for at least two
different antigens selected
from the group consisting of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and
TSPAN8.
In other words, the cancer cell is bound with a ligand comprising at two
different antigen binding
domains wherein a first antigen binding domain is specific for at least one
antigen selected from
the group consisting of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and
TSPAN8 and a
second antigen binding domain is specific for at least one other antigen
selected from the group
consisting of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8.
Preferential
combinations comprise CLA with CD66c and CLA with TSPAN8.
The ligands according to the invention may be used in combination with agents,
which bind to the
antigen and affect the viability of the cancerous cell expressing this
antigen, preferentially kill the
cancerous cell. Examples of such agents are oncolytic viruses, BiTEs , ADCCs
and immunotoxins.
An oncolytic virus is a virus that preferentially infects and kills cancer
cells. As the infected cancer
cells are destroyed by lysis, they release new infectious virus particles to
help destroy the
remaining tumor. Oncolytic viruses are thought not only to cause direct
destruction of the tumor
cells, but also to stimulate host anti-tumor immune responses. Specific
targeting (e.g.
targeting/ligation to the antigens as disclosed) involves modifying the viral
coat proteins to target
Date Recue/Date Received 2022-11-04

18
tumor cells (e.g. with antigen binding domain specific for antigens as
disclosed) while reducing
entry to non-tumor cells.
Bi-specific T-cell engagers (BiTEs ) are a class of artificial bispecific
monoclonal antibodies that
are investigated for the use as anti-cancer drugs. They direct a host's immune
system, more
specifically the T cells' cytotoxic activity, against cancer cells. BiTEs are
fusion proteins consisting
of two single-chain variable fragments (scFvs) of different antibodies, or
amino acid sequences
from four different genes, on a single peptide chain of about 55 kilodaltons.
One of the scFvs binds
to T cells via the CD3 receptor, and the other to a tumor cell via a tumor
specific molecule. Like
other bispecific antibodies, and unlike ordinary monoclonal antibodies, BiTEs
form a link
between T cells and tumor cells. This causes T cells to exert cytotoxic
activity on tumor cells by
producing proteins like perforin and granzymes, independently of the presence
of MHC I or co-
stimulatory molecules. These proteins enter tumor cells and initiate the
cell's apoptosis. This action
mimics physiological processes observed during T cell attacks against tumor
cells.
Antibody-dependent cell-mediated cytotoxicity (ADCC) is a mechanism of attack
by the immune
system that requires the presence of antibodies bound to the surface of target
cells. Antibodies are
formed of a binding region (Fab), which binds to the target antigen and the Fc
region that can be
detected by immune cells via Fc receptors on their surface. These Fc receptors
are found on the
surface of many cells of the immune system, including natural killer cells.
When a natural killer
cell encounter cells coated with antibodies, the Fc regions interact with
their Fc receptors, leading
to the release of perforin and granzyme B. These two chemicals lead to the
tumor cell initiating
programmed cell death (apoptosis). Antibodies known to induce this method of
cell killing include
Rituximab, Ofatuinumab, Trastuzumab, Cetuximab and Alemtuzumab. Third
generation
antibodies that are currently being developed have altered Fc regions that
have higher affinity for
a specific type of Fc receptor, FcyRIIIA, which can increase the rate of ADCC
dramatically.
An immunotoxin is a human-made protein that consists of a targeting portion
linked to a toxin.
When the protein binds to that cell, it is taken in through endocytosis, and
the toxin kills the cell.
These chimeric proteins are usually made of a modified antibody or antibody
fragment, attached
to a fragment of a toxin. The "targeting portion" is composed of the Fv
portion of an antibody that
Date Recue/Date Received 2022-11-04

19
binds specifically to an antigen expressed by a cell, preferably by a specific
cell type. The toxin is
usually a cytotoxic protein derived from a bacterial or plant protein, from
which the natural binding
domain has been removed so that the Fv directs the toxin to the antigen on the
target cell.
pharmaceutical composition
Another object of the invention is a pharmaceutical composition comprising a
population of
engineered cells expressing a CAR as already disclosed, optionally with a
pharmaceutical
acceptable carrier like Composol or NaCl solution.
Use for treatment of cancer
The population of engineered cells as disclosed and/or the pharmaceutical
composition comprising
the population of engineered cells may be used in a method for treatment of
human cancer with
cells expressing the disclosed target molecules, especially of human pancreas
cancer.
The pharmaceutical composition comprises preferable a population of engineered
cells expressing
a CAR as already disclosed herein. In a variant of the invention, the
pharmaceutical composition
is used in combination with a chemotherapeutic, radiation, or immunomodulatory
agent for
treatment of cancer.
The cancer to be treated may include hematopoietic cancer, myelodysplastic
syndrome, pancreatic
cancer, head and neck cancer, cutaneous tumors, minimal residual disease (MRD)
in acute
lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), lung cancer,
breast cancer,
ovarian cancer, prostate cancer, colon cancer, melanoma or other hematological
cancer and solid
tumors, or any combination thereof. In another embodiment, the cancer includes
a hematological
cancer such as leukemia (e.g., chronic lymphocytic leukemia (CLL), acute
lymphocytic leukemia
(ALL), acute myeloid leukemia (AML), or chronic myelogenous leukemia (CML),
lymphoma
(e.g., mantle cell lymphoma, non-Hodgkin's lymphoma or Hodgkin's lymphoma) or
multiple
myeloma, or any combination thereof. Furthermore, the cancer may include an
adult carcinoma
comprising coral and pharynx cancer (tongue, mouth, pharynx, head and neck),
digestive system
cancers (esophagus, stomach, small intestine, colon, rectum, anus, liver,
intrahepatic bile duct,
gallbladder, pancreas), respiratory system cancers (larynx, lung and
bronchus), bones and joint
Date Recue/Date Received 2022-11-04

20
cancers, soft tissue cancers, skin cancers (melanoma, basal and squamous cell
carcinoma),
pediatric tumors (neuroblastoma, rhabdomyosarcoma, osteosarcoma, Ewing's
sarcoma), tumors
of the central nervous system (brain, astrocytoma, glioblastoma, glioma), and
cancers of the breast,
the genital system (uterine cervix, uterine corpus, ovary, vulva, vagina,
prostate, testis, penis,
endometrium), the urinary system (urinary bladder, kidney and renal pelvis,
ureter), the eye and
orbit, the endocrine system (thyroid), and the brain and other nervous system,
or any combination
thereof.
The treatment of cancer may encompass any method which involves at least one
antigen as
disclosed or any combination of antigens as disclosed as target molecule. Such
methods may be
e.g. treatment with agents which bind to the antigen and affect the viability
of the cancerous cell
expressing this antigen, preferentially kill the cancerous cell. Examples are
oncolytic viruses,
BiTEs , ADCCs and immunotoxins as already disclosed.
For the treatment, immune cells, e.g. T cells of a subject may be isolated.
The subject may suffer
from said cancer or may be a healthy subject. These cells are genetically
modified in vitro or in
vivo to express one or more CARs of the invention. These engineered cells may
be activated and
expanded in vitro or in vivo. In a cellular therapy these engineered cells may
be infused to a
recipient in need thereof. These cells may be a pharmaceutical composition
(said cell plus
pharmaceutical acceptable carrier). The infused cells are able to kill (or at
least stop growth of)
cancerous cells expressing one or more of the disclosed antigens in the
recipient. The recipient
may be the same subject from which the cells was obtained (autologous cell
therapy) or may be
from another subject of the same species (allogeneic cell therapy).
In one embodiment of the invention the subject suffering from pancreas cancer
may be treated
with the pharmaceutical composition of the invention together with an
immunomodulatory agent,
such as but not limited to Rapamycin or agents blocking PD-1/PD-L1 or CTLA4
signaling.
In one embodiment of the invention, due to the fact that the cancerous cells
expressing one or more
of the disclosed antigens may be only a subpopulation of the cancerous cells
of the subject the
Date Recue/Date Received 2022-11-04

21
subject may be treated additionally with chemotherapy or radiotherapy.
Chemotherapeutic and
radiation agents suited to treat cancers are well known in the art
In one embodiment of the invention the CAR expressing cells are applied to a
subject suffering
from cancer, especially pancreas cancer as cellular therapy as disclosed above
but in combination
with a second activating CAR, which is also expressed on the same engineered
cells, recognizing
an additional epitope on the cancerous cells expressing one or more of the
disclosed antigens to
increase the specificity of the engineered cells expressing both CARS. This
epitope can be
membrane bound, part of the extracellular matrix, or a soluble component.
In one embodiment of the invention the CAR expressing cells are applied to a
subject suffering
from cancer as cellular therapy as disclosed above but in combination with a
second, inhibitory
CAR, which is also expressed on the same engineered cells, recognizing an
additional epitope to
increase the specificity of the engineered cells expressing both CARS. This
epitope can be
membrane bound, part of the extracellular matrix, or a soluble component.
The immune cells, preferentially T cells engineered to express one or more of
the disclosed
antigens may be administered either alone, or as a pharmaceutical composition
in combination
with diluents and/or with other components such as IL-2 or other cytokines or
cell populations.
Briefly, pharmaceutical compositions of the present invention may comprise a
cell population of
genetically modified cells as described herein, in combination with one or
more pharmaceutically
or physiologically acceptable carriers, diluents or excipients. Such
compositions may comprise
buffers such as neutral buffered saline, phosphate buffered saline and the
like; carbohydrates such
as glucose, mannose, sucrose or dextrans, mannitol; proteins;
polypeptides or amino acids such as glycine; antioxidants; chelating agents
such as EDTA or
glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
Preferentially, the compositions of the present invention are formulated for
intravenous
administration. The administration of cell compositions to the subject may be
carried out in any
convenient manner known in the art.
Date Recue/Date Received 2022-11-04

22
Pharmaceutical compositions of the present invention may be administered in a
manner
appropriate to the disease to be treated. Appropriate dosages may be
determined by clinical trials.
But the quantity and frequency of administration will also be determined and
influenced by such
factors as the condition of the patient, and the type and severity of the
patient's disease.
A pharmaceutical composition comprising the immune cells, preferentially T
cells disclosed herein
may be administered at a dosage of 104 to 109 cells/kg body weight, preferably
105 to 106 cells/kg
body weight. The cell compositions may also be administered several times at
these dosages. The
compositions of cells may be injected directly into a tumor, lymph node, or
site of infection.
Examples
The following examples are intended for a more detailed explanation of the
invention but without
restricting the invention to these examples.
Example 1: Expression of targets on pancreatic cancer
Expression of CLA, CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8 on
xenotransplanted human pancreatic cancer cells indicates the strong abundance
of these markers
on pancreatic cancer cells independent on the tumor microenvironment (Fig 3).
In addition, all
markers are also reproducibly expressed at high levels on primary human
pancreatic cancer cells
(marked Epcarn+) but neither on healthy tumor infiltrating leukocytes (marked
CD45+) cells nor
on other healthy tissue resident cell types (marked Double negative) (Fig 4).
These results were
further validated using immunohistochemistry based detection of target
expression in human
pancreas cancer (Fig 5).
Example 2: Structure of a CAR recognizing pancreas cancer specific targets
The linkers used may comprise an epitope/tag allowing for the detection of the
CAR as shown in
Fig 1. Examples for epitopesitags are YOL, cMYC, or HIS. The pancreas cancer
target specific
binding fragment is derived from one or several antibodies specific for CLA,
CD142, CD73,
CD49c, CD66c, CD104, CD318 and/or TSPAN8. The hinge region may be derived e.g.
from IgG
domains, CD8a, or CD28 and may comprise an epitope/tag allowing for the
detection of the CAR.
The transmembrane domain may be derived e.g. from CD8a or CD28 followed by one
to three
signaling domains. These domains may be derived e.g. from CD28, 4-1BB, 0X40,
or CD3 zeta.
Date Recue/Date Received 2022-11-04

23
Example 3: Structure of dual CAR recognizing pancreas cancer specific targets
Recognition of two or more targets can be solved by either combining multiple
antigen binding
sites on one CAR molecule or by using multiple CAR molecules which are
expressed in one cell
and only work in combination, e.g. by using signaling domains for each of the
CAR constructs
that are inefficient for cell activation when used alone (FIG 2).
Example 4: Amino acid sequence of the CLA specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CLA were as given in SEQ ID
NO:1 and SEQ
ID NO:2, respectively. The relevant sites causing specificity for antigen
binding are the CDRs
according to the IMGT (the international ImMunoGeneTics information system for

immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CLA can be used to
generate a CAR
recognizing CLA. The sequences given in SEQ ID NO:1 and SEQ ID NO:2 are only
exemplary
for sequences which are specific for the antigen CLA (the sequences are given
in one letter code
for amino acids). Other sequences may be used for generating antigen binding
domains of an
antibody or of a CAR which are also specific for the antigen CLA.
Example 5: Amino acid sequence of the CD142 specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CD142 were as given in SEQ
ID NO:3 and SEQ
ID NO:4, respectively. The relevant sites causing specificity for antigen
binding are the CDRs
according to the IMGT (the international ImMunoGeneTics information system for

immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CD142 can be used to
generate a CAR
recognizing CD142. The sequences given in SEQ ID NO:3 and SEQ ID NO:4 are only
exemplary
for sequences which are specific for the antigen CD142 (the sequences are
given in one letter code
for amino acids). Other sequences may be used for generating antigen binding
domains of an
antibody or of a CAR which are also specific for the antigen CD142.
Example 6: Amino acid sequence of the CD73 specific antibody
Date Recue/Date Received 2022-11-04

24
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CD73 were as given in SEQ
ID NO:5 and SEQ
ID NO:6, respectively. The relevant sites causing specificity for antigen
binding are the CDRs
according to the IMGT (the international ImMunoGeneTics information system for

immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CD73 can be used to
generate a CAR
recognizing CD73. The sequences given in SEQ ID NO:5 and SEQ ID NO:6 are only
exemplary
for sequences which are specific for the antigen CD73 (the sequences are given
in one letter code
for amino acids). Other sequences may be used for generating antigen binding
domains of an
antibody or of a CAR which are also specific for the antigen CD73.
Example 7: Amino acid sequence of the CD49c specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CD49c were as given in SEQ
ID NO:7 and SEQ
ID NO:8, respectively. The relevant sites causing specificity for antigen
binding are the CDRs
according to the IMGT (the international ImMunoGeneTics information system for

immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CD49c can be used to
generate a CAR
recognizing CD49c. The sequences given in SEQ ID NO:7 and SEQ ID NO:8 are only
exemplary
for sequences which are specific for the antigen CD49c (the sequences are
given in one letter code
for amino acids). Other sequences may be used for generating antigen binding
domains of an
antibody or of a CAR which are also specific for the antigen CD49c.
Example 8: Amino acid sequence of the CD66c specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CD66c were as given in SEQ
ID NO:9 and SEQ
ID NO:10, respectively. The relevant sites causing specificity for antigen
binding are the CDRs
according to the MGT (the international ImMunoGeneTics information system for
immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CD66c can be used to
generate a CAR
recognizing CD66c. The sequences given in SEQ ID NO:9 and SEQ ID NO:10 are
only exemplary
Date Recue/Date Received 2022-11-04

25
for sequences which are specific for the antigen CD66c (the sequences are
given in one letter code
for amino acids). Other sequences may be used for generating antigen binding
domains of an
antibody or of a CAR which are also specific for the antigen CD66c.
Example 9: Amino acid sequence of the CD104 specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CD104 were as given in SEQ
ID NO:11 and
SEQ ID NO:12, respectively. The relevant sites causing specificity for antigen
binding are the
CDRs according to the IMGT (the international ItnMunoGeneTics information
system for
immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CD104 can be used to
generate a CAR
recognizing CD104. The sequences given in SEQ ID NO:!! and SEQ ID NO:12 are
only
exemplary for sequences which are specific for the antigen CD104 (the
sequences are given in one
letter code for amino acids). Other sequences may be used for generating
antigen binding domains
of an antibody or of a CAR which are also specific for the antigen CD104.
Example 10: Amino acid sequence of the CD318 specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to CD318 were as given in SEQ
ID NO:13 and
SEQ ID NO:14, respectively. The relevant sites causing specificity for antigen
binding are the
CDRs according to the IMGT (the international ImMunoGeneTics information
system for
immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for CD318 can be used to
generate a CAR
recognizing CD318. The sequences given in SEQ ID NO:13 and SEQ ID NO:14 are
only
exemplary for sequences which are specific for the antigen CD318 (the
sequences are given in one
letter code for amino acids). Other sequences may be used for generating
antigen binding domains
of an antibody or of a CAR which are also specific for the antigen CD318.
Example 11: Amino acid sequence of the TSPAN8 specific antibody
The amino acid sequences of the variable portions of the immunoglobulin heavy
chain and light
chain of the used antibody specifically binding to TSPAN8 were as given in SEQ
ID NO:15 and
Date Recue/Date Received 2022-11-04

26
SEQ ID NO:16, respectively. The relevant sites causing specificity for antigen
binding are the
CDRs according to the IMGT (the international ImMunoGeneTics information
system for
immunoglobulins or antibodies) definition which are underlined in the
sequence. These sequences
or any sequences derived thereof with a specificity for TSPAN8 can be used to
generate a CAR
recognizing TSPAN8. The sequences given in SEQ ID NO:15 and SEQ ID NO:16 are
only
exemplary for sequences which are specific for the antigen TSPAN8 (the
sequences are given in
one letter code for amino acids). Other sequences may be used for generating
antigen binding
domains of an antibody or of a CAR which are also specific for the antigen
TSPAN8.
Example 12: Validation of CAR expression
75.000 HEK 293T cells were inoculated in an 48-well. Cells were transfected
with 0.5 Lig plasmid
using the MACSfectinTM transfection protocol 24 h later. Cells were detached
48 h post
transfection using PBS supplemented with 1 mM EDTA. One third of the cells
transfected with
one plasmid was stained with an Anti-CD271 antibody (Miltenyi Biotec GmbH)
following the
recommended protocol. Another third was incubated 30 min at 4 C with an Anti-
Mouse IgG (Fab
specific) antibody produced in goat (Sigma Aldrich) with 10 ig/m1 antibody
concentration. After
a washing step cells were incubated with an Anti-Goat IgG (H+L) Cross-Adsorbed
Secondary
Antibody produced in chicken (Thermo Fisher Scientific) for 30 min at 4 C
with 10 ps/m1
antibody concentration. The last third was stained solely as described with
the secondary antibody
as a background control. Samples were measured using a MACSQuantTM 10
analyzer. Fig. 6 A
shows the percentage of cells expressing LNGFR on the surface as a readout of
transfection
efficacy and construct expression. Fig. 6 B shows the percentage of cells
expressing the CAR
construct on the surface. With few exceptions, all CAR molecules could be
successfully expressed.
Example 13: Generation of lentiviral expression vectors
The pancreas cancer specific CARs were cloned into third generation SIN-
lentiviral vector
constructs under the control of the human PGK promoter. Transient transfection
of HEK 293T
cells with this expression plasmid and further plasmids encoding the
structural proteins gag-pol,
rev and VSV-G envelope protein resulted in the release of viral vector
particles into the
supernatant. The viral vector particles were subsequently enriched by low
speed centrifugation and
stored at -70 C.
Date Recue/Date Received 2022-11-04

27
Example 14: T cell separation and genetic modification with pancreas cancer
specific CARS
Primary T cells were isolated from donor apheresis or buffy coat samples using
MicroBeadsTm and
MACS technology (Miltenyi Biotec GmbH, Germany) to reach purities of over 90%
(CD3+
cells). Magnetically enriched cells were washed and resuspended in TexMACSTm
medium
supplemented with 200 IU/mL human recombinant 1L-2 (Miltenyi Biotec GmbH,
Germany). The
T cells were then stimulated by addition of the GMP TransActTm CD3/CD28
Reagent (Miltenyi
Biotec GmbH, Germany).
After 24 hours, successful T cell stimulation was confirmed by staining the T
cells with CD25 and
CD69 antibodies and analysis by flow cytometry in a MACSQuantTm Analyzer
(Miltenyi Biotec
GmbH, Germany). The stimulated T cells were then transduced by adding
lentiviral vectors
encoding pancreas cancer specific CARS at an MOI = 0.5-2. After 4 days of
static culture the cells
were washed to remove excess viral vector and TransActTm Reagent and were
cultivated for a
further 5-10 days. The efficiency of viral transduction was measured by
staining the surface
expression of pancreas cancer specific CARS among live CD3+ cells using anti-
human Fc
fluorochrome and flow cytometry. The number of gene marked T cells ranged
between 10 and
60%, depending on the MOI used.
Example 15: Pancreas cancer specific CARs functionality
Cells expressing one or more of the pancreas cancer specific targets or cells
not expressing these
targets were incubated for 5 or 24 hours with expanded T cells expressing
pancreas cancer specific
CARS or, as a control, with non-transduced T cells at varied effector to
target cell ratios. Specific
target cell killing was analyzed by flow cytometry.Altematively, the effector
cells were
restimulated with cell lines which were target-positive or -negative. Cytokine
production (IFN-y,
IL-2, TNF-a) as well as degranulation (CD107a) were analyzed by flow
cytometry. Only T cells
carrying the pancreas cancer specific CARS were able to kill the target cells,
showed increased
cytokine production as well as degranulation marker upregulation.
Furthermore, the killing efficacy and kinetics were analyzed by long-term co-
culture of CAR T
cells and pancreatic cancer cells. T cells were isolated from a whole blood
donation of a healthy
donor using the Pan T Cell Isolation Kit (Miltenyi Biotec, Germany). Isolated
T cells were
Date Recue/Date Received 2022-11-04

28
activated in TexMACSTm GMP medium (Miltenyi Biotec, Germany) supplemented with
40 IU/m1
IL-2 (Miltenyi Biotec, Germany) using MACS GMP T Cell TransActTm (Miltenyi
Biotec,
Germany). After 24 h T cells were transduced with lentiviral vectors
containing the CAR
constructs at an MOI of 2. Subsequently, T cells were cultivated in TexMACSTm
GMP medium
supplemented with 40 1U/m1 IL-2. On day 12 post transduction CAR expression
was assessed via
flow cytometric measurement of the reporter protein LNGFR. CAR positive T
cells were adjusted
to same numbers and inoculated in 96 well plates with pancreatic
adenocarcinoma cell line BxPC3
in 200 Ill TexMACSTm GMP medium. Prior to this assay, BxPC3 cells were
transduced with
lentiviral vectors containing GFP. GFP positive cells were tracked and
analyzed using the
IncuCyteTm S3 (Essen BioScience, Germany). The "Green Object confluence" was
used as a read
out as it inversely correlates with the specific lysis of GFP expressing
adherent target cells. All
CAR constructs mediated efficient killing of tumor cells as compared to the
mock control. Results
are shown in Fig.7.
Example 16: Amino acid sequences of CAR binding domains recognizing CLA
For the antigen binding domain of a CAR specifically recognizing CLA, scFv's
were used having
the amino acid sequences of either SEQ ID NO:17 or SEQ ID NO:18 (the sequences
are given in
one letter code of amino acids).
Example 17: Amino acid sequences of CAR binding domains recognizing CD142
For the antigen binding domain of a CAR specifically recognizing CD142, scFv's
were used
having the amino acid sequences of either SEQ ID NO:19 or SEQ NO:20 (the
sequences are
given in one letter code of amino acids).
Example 18: Amino acid sequences of CAR binding domains recognizing CD73
For the antigen binding domain of a CAR specifically recognizing GD73, scFv's
were used having
the amino acid sequences of either SEQ ID NO:21 or SEQ ID NO:22 (the sequences
are given in
one letter code of amino acids).
Example 19: Amino acid sequences of CAR binding domains recognizing CD49c
Date Recue/Date Received 2022-11-04

29
For the antigen binding domain of a CAR specifically recognizing CD49c, scFv's
were used
having the amino acid sequences of either SEQ ID NO:23 or SEQ ID NO:24 (the
sequences are
given in one letter code of amino acids).
Example 20: Amino acid sequences of CAR binding domains recognizing CD66c
For the antigen binding domain of a CAR specifically recognizing CD66c, scFv's
were used
having the amino acid sequences of either SEQ ID NO:25 or SEQ ID NO:26 (the
sequences are
given in one letter code of amino acids).
Example 21: Amino acid sequences of CAR binding domains recognizing CD104
For the antigen binding domain of a CAR specifically recognizing CD104, scFv's
were used
having the amino acid sequences of either SEQ ID NO:27 or SEQ ID NO:28 (the
sequences are
given in one letter code of amino acids).
Example 22: Amino acid sequences of CAR binding domains recognizing CD318
For the antigen binding domain of a CAR specifically recognizing CD318, scFv's
were used
having the amino acid sequences of either SEQ ID NO:29 or SEQ ID NO:30 (the
sequences are
given in one letter code of amino acids).
Example 23: Amino acid sequences of CAR binding domains recognizing TSPAN8
For the antigen binding domain of a CAR specifically recognizing TSPAN8,
scFv's were used
having the amino acid sequences of either SEQ ID NO:31 or SEQ ID NO:32 (the
sequences are
given in one letter code of amino acids).
Example 24: Co-expression of CLA and CD142, CD73, CD49c, CD66c, CD104, CD318
and
TSPAN8 on primary human pancreatic cancer cells
Human pancreas adenocarcinoma tissue was dissociated, stained and analyzed.
Fig 8A shows the
general gating strategy: after debris exclusion dead cells were excluded with
a propidium iodide
staining. Unwanted doublets were excluded by plotting the height against the
area for forward
Date recue/Date received 2023-05-05

30
scatter. EpCAM+, CD45+ as well as CD45-/EpCAM- cells were discriminated and
further
analyzed with PE or APC conjugated antibodies specific for the targets. Fig 8B
shows co-
expression of CLA and CD142, CD73, CD49c, CD66c, CD104, CD318 and TSPAN8 on
primary
human pancreatic cancer cells. The results indicate a strong co-expression
among these markers
which is a pre-requisite for dual targeting (Fig. 8 A and B).
Sequences
SEQ ID NO:1
CLA VH
EVQLVESGGGLVQP GN SLKL SC S ASGFTF S SY GMHW IRQAP GEGL DWVAY IS SS S GTVY
ADAVKARFTISRDNAKNTLYLQLNSLKSEDTAIYYCARAQNWDLFDYWGQGVMVTVS
SEQ ID NO:2
CLA VL
Q1MLTQQAESLWISPGERVSITCRASQSLLYTDGKHYLSWYQQKPGQTTKALIYHASVR
TD GVPTRF IGS GS GTEFTL SIEHVQPEDFAIYYCLQ TLK SPFTF GSGTKLEIK
SEQ ID NO:3
CD142 VH
QVQLKQSGPGLVQPSQSLSITCTVSGF SLSNYGVHWVRQSPGKGLEWLGVIWSGGSTDY
NVAFISRLITTKDNSKSQVFLKMNSLQADDTAIYFCARTTGSVFNAMDHWGQGTSVTVS
SEQ ID NO:4
CD142 VL
QIVLTQ SPALM SA SP GEKVTMTC SAS SSVTYMYWY QQKPR SSPKPWIYLT SNLA SGVPA
RF SGSGSGTSYSLTISSVEAEDAATYYCQQW SSNPLTFGAGTKLELK
SEQ ID NO:5
CD73 VH
Date recue/Date received 2023-05-05

31
EVQL QQ SGAELVKP GA SVKL SCTASGFNIKDTYIHWVKQRPEQGLEWIGRIDPATGNTE
YDPKFQGKATITADTS SNTAYLHLSSL TS ED TAVYYC ARGYY GS SYPPWFAYWGQGTL
VTVSA
SEQ ID NO:6
CD73 VL
DIVMTQSHKFMSTSVGDRVSITCKASQDVG SAVAWYQQKPGQSPICLLIYWASTRHTGV
PDRFT G S GS GTDFTLTI SNVQ SEDLADYFCQQYS SYPLTFGAGTKLELK
SEQ ID NO:7
CD49c VH
EVQL QQ SGAELVKP GA SVKLSCTASGFNIKDTYMHWVKQRPEQGLEWIGRIDPANGHT
KYDPKF QGKATITADTSSNAAYL QLN SLTSEDTAVYY CARRVA YAMDYW GQGTS VT V
SS
SEQ ID NO:8
CD49c VL
ENVL TQ SP AIM SA SP GEKVTMTC SASS SVTYMHWYQQKSSTSPKLWIYDTSKLAS GVPG
RF SGSGSGN SY SL TI S SM EAEDVATYC CF QGSGYPLTF GrGGTICLEIK
SEQ ID NO:9
CD66c VH
QVTLKESGPGILKPSQTLSLTCSF S GF S L S TS GMGVGWIRQP S GK S LEW LAH IW WND ER
YYNPSLICNQLTISKDTSRNQVFLKITSVDTADTATYYCARSPRGYFDYWGHGTTLTVS S
SEQ ID NO:10
CD66c VL
DIVMTQSQKFMSTSVGDRVSVTCKAS QNVVTNVAWYQQTPGQSPKALIYSASYRYSGV
PDRF S GS GS GTDF TLTI SNVQSGDLAEYFC QOYNSYPLTFGAGTKLELK
SEQ ID NO:11
Date Regue/Date Received 2022-11-04

32
CD1O4VH
QVNLLQSGAALVKPGASVKLSCKASGYTFTDYYIFWVKQSHGKSLEWIGYINPNSGSTN
YNEKFKRKATLSVDKSTNTAYMELSRLTSEDSATYYCTRRAYYGYNPFDYWGQGVMV
TVSS
SEQ ID NO:12
CD1O4VL
DIQMTQTP SSMPASLGERVTISCRASRGINNYLSWYQQNLDGTIKPLIYYTSNLQSGVP S
RF SGSGSGTDYSLTISSLEPEDFAMYYCOOYDSSPWTF GGGTKLELK
SEQ ID NO:13
CD318 NTH
EVQL QQ SGAELVRPGALVKL SCKASGFNIKDYYIHWVKQRPEQ GLEWIGWIDPENGHTI
YDPKFQGKASITADTSSNTAYLQLSSLTSEDTAVYYCARLTGTTYAMDYWGQGTSVTV
SS
SEQ ID NO:14
CD318 VL
DIVMTQSHKFMSTSVGDRVSITCKAS QDVSTAVAWYQQKSGQSPICLLIYWASTRHTGV
PDRFTGSGSGTDYTLTISSVQAEDLALYYCQQHYSTPYTFGGGTKLEIK
SEQ ID NO:15
TSPAN8 VH
EVICLLESGGGLVQPGGSMRLSCAASGFTFTDFYMNWIRQPAGKAPEWLGFIRNKASGY
TTEYNPSVKGRFTISRDNTQNMLYLQMNTLRAEDTATYYCARAHSYYGYDYFDYWGQ
GVMVTVSS
SEQ ID NO:16
TSPAN8 YL
DIQMTQSPASLSASLEEIVTITCQASQDIGNWLSWYQQKPGKSPQLLIYGATSLADGVPS
RF SGSRSGTQYSLKISRLQVEDIRIYYC LOAY SAPWTF GGGTKLELK
Date Regue/Date Received 2022-11-04

33
SEQ ID NO:17
CLA specific scFv VH-linker-VL
EVQLVESGGGLVQPGN SLKL SC SA SGF TF SSYGMHWIRQAPGEGLDWVAYIS SS SGTVY
ADAVKARF TISRDNAKNTLYLQLNSLKSEDTAIYYCARAQNWDLFDYWGQGVMVTVS
SGGGGSGGGGSGGGGSQIMLTQQAESLWISPGERVSITCRA SQSLLYTDGKHYLSWYQQ
KPGQTTKALIYHASVRTDGVPTRFIGSGSGTEFTLSIEHVQPEDFAIYYCLQTLKSPFTFGS
GTKLEIK
SEQ ID NO:18
CLA specific scFv VL-linker-VH
QIMLTQQAESLWISPGERVSITCRASQ SLLYTDGKHYL S WYQQKPGQTTKALIYHASVR
TDGVPTRFIGSGSGTEFTLSIEHVQPEDFAIYYCLQTLKSPFTFGSGTKLEIKGGGGSGGG
GSGGGGSEVQLVESGGGLVQPGNSLKLSCSASGFTF S SY GMHWIRQAPGEGLDWVAYI
SSSSGTVYADAVKARFTISRDNAKNTLYLQLNSLKSEDTAIYYCARAQNWDLFDYWGQ
GVMVTVSS
SEQ ID NO:19
CD142 specific CAR sequence VH-linker-VL
QVQLKQSGPGLVQPSQSLSITCTVSGF SLSNYGVHWVRQSPGKGLEWLGVIWSGGSTDY
NVAFISRLIITKDNSKSQVFLKMNSLQADDTAIYFCARTTGSVFNAMDHWGQGTSVTVS
SGGGGSGGGGSGGGGSQIVLTQ SPALMSA SPGEKVTMTC SA SS SVTYMYWYQQKPRS S
PKPWIYLTSNLASGVPARF SGSGSGTS YSLTISSVEAEDAATYYCQQW S SNP LTF GAGTK
LELK
SEQ ID NO:20
CD142 specific CAR sequence VL-linker-VH
QIVLTQ SPA LMSA SPGEKVTMTC SAS S SVTYMYWYQQKPRSSPKPWIYLTSNLASGVPA
RF SG SGS GT SY SLTIS SVEAEDAATYY CQQW SSNPLTF GAGTKLELKGGGGS GGGG SGG
GGSQVQLKQS GP GLVQPS Q SLS ITCTVSGF SLSNYGVHWVRQSP GKGLEWLGVIWSGGS
Date Regue/Date Received 2022-11-04

34
TDYNVAFI SRLIITKDN SKS QVFLKMNSLQADDTAIYF CARTTGSVFNAMDHWGQGTSV
TVSS
SEQ ID NO:21
CD73 specific CAR sequence VH-linker-VL
EVQLQQSGAELVKPGASVKLSCTASGFNIKDTYIHWVKQRPEQGLEWIGRIDPATGNTE
YDPKF Q GKATITADT S SNTAYLHL S SL TS ED TAVYYC ARGYYGS SYPPWFAYWGQGTL
VTVSAGGGGSGGGGSGGGGSDIVMTQSHKFMSTSVGDRVSITCKASQDVGSAVAWYQ
QKPGQSPKWYWASTRHTGVPDRFTGSGSGTDFTLTISNVQSEDLADYFCQQYSSYPLT
FGAGTKLELK
SEQ ID NO:22
CD73 specific CAR sequence VL-linker-VH
D1VMTQSHKFMSTSVGDRVSITCKASQDVGSAVAWYQQKPGQ SPKLLIY WASTRHTGV
PDRFT GS G S GTDFTLTISNVQ SEDLAD YF C QQY S SYP L TF GA GTKL EL KGGG G SGG GGS

GGGGSEVQLQQ SGAELVKPGASVKLSCTASGFNIKDTYIHWVKQRPEQGLEWIGRIDPA
TGNTEYDPKFQGKATITADTSSNTAYLHLSSLTSEDTAVYYCARGYYGSSYPPWFAYW
GQGTLVTVSA
SEQ ID NO:23
CD49c specific CAR sequence VH-linker-VL
EVQLQQSGAELVKPGA SVKLSCTASGFNIKDTYMHIATVKQRPEQGLEWIGRIDPANGHT
KYDPKFQGKATITADTSSNAAY LQLNSLTSEDTAVYYCARRVAYAMDYWGQGTSVTV
S S GG GGS GGGGS GGGG SENVLT Q SPA IM SA SPGEKVTMTC SA S S SVTYMHWYQQKS S T
SPKLWIYDTSKLASGVPGRFSGS GS GNSYSL TIS SMEAEDVATYC CFQGSGYPLTF GGGT
KLEIK
SEQ ID NO:24
CD49c specific CAR sequence VL-linker-VH
ENVL TQ SPAIM SA SP GEKVTMTC SASS SVTYMHWYQQKSSTSPKLWIYDTSKLAS GVPG
RF SGSGSGN SYSLTISSMEAEDVATYC CFQGSGYP L _________________________________
GGGTKLEIKGGGGSGGGGSGG
Date Regue/Date Received 2022-11-04

35
GGSEVQLQQSGAELVKPGASVKLSCTASGFNIKDTYMHWVKQRPEQGLEWIGRIDPAN
GHTKYDPKFQGKATITADTS SNAAYL QLNSLTSEDTAVYYCARRVAYAMDYWGQGTS
VTVSS
SEQ ID NO:25
CD66c specific CAR sequence VH-linker-VL
QVTLKESGPGILKPS QTLSLTC SF SGFSLSTSGMGVGWIRQP SGKSLEWLAHIWWNDER
YYNP SLKNQLTIS KDTSRN QVF LKITSVDTADTATYYCARSPRGYFDYWGH Gilt TVS S
GGGGSGGGGSGGGGSDIVMTQSQKFMSTSVGDRVSVTCKASQNVVTNVAWYQQTPG
Q SPKALIY S ASYRYSGVPDRF SG SGSGTDFTLTISNVQ SGDL AEYF CQQYNSYPLTF GAG
TKLELK
SEQ ID NO:26
CD66c specific CAR sequence VL-linker-VH
DIVMTQSQKFMSTSVGDRVSVTCKAS QNVVTNVAWYQQTPGQSPKALIYSASYRYSGV
PDRF S GS GS GTDF TLTI SNVQSGDLAEYF C QQYNSYPLTFGAGTKLELKGGGGSGGGG S
GGGGSQVTLKESGPGILKPSQTL SLTC SF SGF SL STSGMGVGWIRQPSGKSLEWLAHIW
WNDERYYNPSLKNQLTISKDTSRNQVFLKITSVDTADTATYYCARSPRGYFDYWGHGT
TLTVSS
SEQ ID NO:27
CD104 specific CAR sequence VH-linker-VL
QVNL LQ SGAALVKPGASVKL SC KAS GYTF TDYYIFWVKQSHGKS LEWIGY INPNSGSTN
YNEKFKRKATLSVDKSTNTAYMELSRLTSEDSATYYCTRRAYYGYNPFDYWGQGVMV
TVSSGGGGSGGGGSGGGGSDIQMTQTPSSMPASLGERVTISCRASRGINNYL SWYQQNL
DGTIKPLIYYTSNLQSGVPSRFSGSGSGTDY SLTIS SL EP EDFAMYY CQ QYD S SPWTF GGG
TKLELK
SEQ ID NO:28
CD104 specific CAR sequence VL-linker-VH
Date Recue/Date Received 2022-11-04

36
DIQMTQTP SSMPASLGERVTISCRASRGINNYLSWYQQNLDGTIKPLIYYTSNLQSGVP S
SGSGSGTDY SL TIS SUP EDFAMYYCQQYDSSPWTF GGGTKLELKGGGGSGGGGSGG
GGSQVNLLQSGAALVKPGASVKLSCKASGYTFTDYYIFWVKQSHGKSLEWIGYINPNSG
STNYNEKFKRKATLSVDKSTNTAYMELSRLTSEDSATYYCTRRAYYGYNPFDYWGQG
VMVTVSS
SEQ ID NO:29
CD318 specific CAR sequence VH-linker-VL
EVQL QQ SGAELVRPGA LVKL SCKA SGFNIKDYYIHWVKQRPEQGLEWIGWIDPENGHTI
YDPKFQGKASITADTSSNTAYLQ LSSLTSEDTAVYYCARLTGTTYAMDYWGQGTSVTV
SSGGGGSGGGGSGGGGSDIVMTQ SHKFM STSVGDRVSITCKAS QDVSTAVAWYQ QK SG
QSPICLLIYWASTRHTGVPDRFTGSGSGMYTLTISSVQAEDLALYYCQQHYSTPYTEGG
GTKL EIK
SEQ ID NO:30
CD318 specific CAR sequence VL-linker-VH
DIVMTQ SHKF MS TSVGDRVS ITCKAS QDVS TAVA WY Q QK SGQSPKLLII(WA STRHTGV
PDRFTGSGSG _______________________________________________________________ ID
YTLTI SSVQAEDLA L YYC QQHY STPYTFGGGTKLEIKGGGGSGGGGS
GGGGSEVQLQQ S GAELVRP GALVKL S CKA SGF NIKDY YIHW VKQRPEQ GL EWIGWIDP
ENGHTIYDPKFQ GKASITADTSSNTAYLQLS SLTSEDTAVYYCARLTGTTYAMDYWGQ
GTSVTVSS
SEQ ID NO:31
TSPAN8 specific CAR sequence VH-linker-VL
EVKLLESGGGLVQPGGSMRLSCAASGFTFTDFYMNWIRQPAGKAPEWLGFIRNKASGY
TTEYNP SVKGRF TISRDNTQNMLYLQMNTLRAEDTATYYCARAHSYYGYDYFDYWGQ
GVMVTVSSGGGGSGGGGSGGGGSDIQMTQ SPA SL SA SLEEIVTITCQA SQD IGNWLSWY
QQKP GKSP QLL IY GAT SLADGVP SRFSGSRS GTQYSLKISRLQVEDIRIYYCLQAYSAPW
TFGGGTKLELK
Date Regue/Date Received 2022-11-04

37
SEQ ID NO:32
TSPAN8 specific CAR sequence VL-linker-VH
DIQMTQSPASLSASLEEIVTITCQASQDIGNWLSWYQQKPGKSPQLLIYGATSLADGVPS
RF SGSRSGTQYSLKISRLQVEDIRTYYCLQAYSAPWTFGGGTKLELKGGGGSGGGGSGG
GGSEVKLLESGGGLVQPGGSMRLSCAASGFTFTDFYMNWIRQPAGKAPEWLGFIRNKA
SGYTTEYNPSVKGRFTISRDNTQNMLYLQMNTLRAEDTATYYCARAHSYYGYDYFDY
WGQGVMV'TVSS
Date recue/Date received 2023-05-05

Representative Drawing

Sorry, the representative drawing for patent document number 2982963 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2024-02-06
(22) Filed 2017-10-19
(41) Open to Public Inspection 2018-04-20
Examination Requested 2022-06-23
(45) Issued 2024-02-06

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $210.51 was received on 2023-10-09


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-10-21 $100.00
Next Payment if standard fee 2024-10-21 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2017-10-19
Maintenance Fee - Application - New Act 2 2019-10-21 $100.00 2019-09-24
Registration of a document - section 124 2019-10-21 $100.00 2019-10-21
Maintenance Fee - Application - New Act 3 2020-10-19 $100.00 2020-09-23
Maintenance Fee - Application - New Act 4 2021-10-19 $100.00 2021-10-11
Advance an application for a patent out of its routine order 2022-06-23 $508.98 2022-06-23
Request for Examination 2022-10-19 $814.37 2022-06-23
Maintenance Fee - Application - New Act 5 2022-10-19 $203.59 2022-10-10
Maintenance Fee - Application - New Act 6 2023-10-19 $210.51 2023-10-09
Final Fee $306.00 2023-12-20
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
MILTENYI BIOTEC B.V. & CO. KG
Past Owners on Record
MILTENYI BIOTEC GMBH
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Request for Examination 2022-06-23 3 75
Special Order / Amendment 2022-06-23 8 228
Claims 2022-06-23 3 119
Acknowledgement of Grant of Special Order 2022-07-19 1 205
Examiner Requisition 2022-07-22 4 258
Amendment 2022-11-04 55 2,563
Drawings 2022-11-04 8 370
Description 2022-11-04 37 2,590
Claims 2022-11-04 1 35
Examiner Requisition 2023-02-20 7 386
Amendment 2023-05-05 14 466
Claims 2023-05-05 1 29
Description 2023-05-05 37 2,544
Abstract 2017-10-19 1 10
Description 2017-10-19 37 1,788
Claims 2017-10-19 2 68
Drawings 2017-10-19 8 245
Cover Page 2018-03-28 1 27
Final Fee 2023-12-20 4 107
Cover Page 2024-01-08 2 31
Electronic Grant Certificate 2024-02-06 1 2,528
Examiner Requisition 2023-06-14 7 447
Amendment 2023-10-13 14 805
Claims 2023-10-13 1 24

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :